Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Am J Trop Med Hyg ; 95(5): 1148-1156, 2016 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-27645784

RESUMO

Passive immunization with anti-dengue virus (DENV) immune serum globulin (ISG) or monoclonal antibodies (Mabs) may serve to supplement or replace vaccination for short-term dengue immune prophylaxis. In the present study, we sought to establish proof-of-concept by evaluating several DENV-neutralizing antibodies for their ability to protect rhesus macaques against viremia following live virus challenge, including human anti-dengue ISG, and a human Mab (Mab11/wt) and its genetically engineered variant (Mab11/mutFc) that is unable to bind to cells with Fc gamma receptors (FcγR) and potentiate antibody-dependent enhancement (ADE). In the first experiment, groups of animals received ISG or Mab11/wt at low doses (3-10 mg/kg) or a saline control followed by challenge with DENV-2 at day 10 or 30. After passive immunization, only low-titered circulating virus-neutralizing antibody titers were measured in both groups, which were undetectable by day 30. After challenge at day 10, a reduction in viremia duration compared with the control was seen only in the ISG group (75%). However, after a day 30 challenge, no reduction in viremia was observed in both immunized groups. In a second experiment to test the effect of higher antibody doses on short-term protection, groups received either ISG, Mab11/wt, Mab11/mutFc (each at 25 mg/kg) or saline followed by challenge with DENV-2 on day 10. Increased virus-neutralizing antibody titers were detected in all groups at day 5 postinjection, with geometric mean titers (GMTs) of 464 (ISG), 313 (Mab11/wt), and 309 (Mab11/mutFc). After challenge, there was complete protection against viremia in the group that received ISG, and a reduction in viremia duration of 89% and 83% in groups that received Mab11/wt and Mab11/mutFc, respectively. An in vitro ADE assay in Fcγ receptor-bearing K562 cells with sera collected immediately before challenge showed increased DENV-2 infection levels in the presence of both ISG and Mab11/wt, which peaked at a serum dilution of 1:90, but not in Mab11/mutFc containing sera. The results suggest that antibody prophylaxis for dengue might be beneficial in eliminating or reducing viral loads thereby minimizing disease progression. Our results also suggest that blocking FcγR interactions through Mab11 Fc engineering may further prevent ADE.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/sangue , Dengue/prevenção & controle , Imunização Passiva , Soroglobulinas/uso terapêutico , Animais , Anticorpos Facilitadores , Dengue/sangue , Vírus da Dengue , Modelos Animais de Doenças , Humanos , Imunoglobulina G/uso terapêutico , Células K562 , Macaca mulatta/sangue , Macaca mulatta/imunologia , Macaca mulatta/virologia , Receptores de IgG/imunologia , Carga Viral , Viremia/sangue , Viremia/prevenção & controle
2.
PLoS One ; 11(4): e0152811, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27035715

RESUMO

Dengue virus purified inactivated vaccines (PIV) are highly immunogenic and protective over the short term, but may be poor at inducing cell-mediated immune responses and long-term protection. The dengue nonstructural protein 3 (NS3) is considered the main target for T-cell responses during viral infection. The amino (N)-terminal protease and the carboxy (C)-terminal helicase domains of DENV-2 NS3 were expressed in E. coli and analyzed for their immune-potentiating capacity. Mice were immunized with DENV-2 PIV with and without recombinant NS3 protease or NS3 helicase proteins, and NS3 proteins alone on days 0, 14 and 28. The NS3 helicase but not the NS3 protease was effective in inducing T-cell responses quantified by IFN-γ ELISPOT. In addition, markedly increased total IgG antibody titer against virus antigen was seen in mice immunized with the PIV/NS3 helicase combination in the ELISA, as well as increased neutralizing antibody titer measured by the plaque reduction neutralization test. These results indicate the potential immunogenic properties of the NS3 helicase protein and its use in a dengue vaccine formulation.


Assuntos
Anticorpos Antivirais/biossíntese , Linfócitos T CD4-Positivos/imunologia , Vírus da Dengue/imunologia , Recombinação Genética , Vacinas de Produtos Inativados/imunologia , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Interferon gama/biossíntese , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos DBA , RNA Helicases/imunologia , Serina Endopeptidases/imunologia
3.
Am J Trop Med Hyg ; 94(6): 1348-1358, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27022153

RESUMO

We evaluated the safety and immunogenicity of two doses of a live-attenuated, tetravalent dengue virus vaccine (F17/Pre formulation) and a booster dose in a dengue endemic setting in two studies. Seven children (7- to 8-year-olds) were followed for 1 year after dose 2 and then given a booster dose (F17/Pre formulation), and followed for four more years (Child study). In the Infant study, 49 2-year-olds, vaccinated as infants, were followed for approximately 3.5 years after dose 2 and then given a booster dose (F17) and followed for one additional year. Two clinically notable events were observed, both in dengue vaccine recipients in the Infant study: 1 case of dengue approximately 2.7 years after dose 2 and 1 case of suspected dengue after booster vaccinations. The booster vaccinations had a favorable safety profile in terms of reactogenicity and adverse events reported during the 1-month follow-up periods. No vaccine-related serious adverse events were reported during the studies. Neutralizing antibodies against dengue viruses 1-4 waned during the 1-3 years before boosting, which elicited a short-lived booster response but did not provide a long-lived, multivalent antibody response in most subjects. Overall, this candidate vaccine did not elicit a durable humoral immune response.


Assuntos
Vacinas contra Dengue/normas , Dengue/prevenção & controle , Esquemas de Imunização , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Criança , Pré-Escolar , Dengue/epidemiologia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/imunologia , Humanos , Imunização Secundária , Lactente , Tailândia/epidemiologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/normas
4.
Am J Trop Med Hyg ; 93(5): 1052-4, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26240157

RESUMO

Japanese encephalitis virus (JEV) is endemic in the Republic of Korea (ROK), posing a medical threat to more than 29,000 U.S. Forces military personnel currently deployed in the ROK. The objective of this study was to provide data on the risk of JEV exposure among U.S. Forces in the ROK. One thousand U.S. Army Soldiers were randomly selected for the study from the cohort of infantry Soldiers deployed in the ROK for a period of at least 330 days from 2008 to 2011. Pre- and post-deployment serum specimens were tested for the presence of JEV antibodies by plaque reduction neutralization test. A total of 2/1,000 (0.2%) U.S. Army Soldiers post-deployment specimens tested positive for JEV antibody. Results from the pre-deployment specimens indicated one true seroconversion and one with titers suggestive of a JEV infection. These results indicate a low, but nonzero risk of JEV exposure among U.S. Army Soldiers in the ROK.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/epidemiologia , Militares/estatística & dados numéricos , Adolescente , Adulto , Animais , Estudos de Coortes , Encefalite Japonesa/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , República da Coreia/epidemiologia , Soroconversão , Estados Unidos , Adulto Jovem
5.
Lancet Infect Dis ; 15(5): 519-27, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25739878

RESUMO

BACKGROUND: Chikungunya is an emerging arthropod-borne disease that has spread from tropical endemic areas to more temperate climates of the USA and Europe. However, no specific treatment or preventive measure is yet available. We aimed to investigate the immunogenicity and safety of a live recombinant measles-virus-based chikungunya vaccine. METHODS: We did a randomised, double-blind, placebo-controlled, active-comparator, phase 1, dose-escalation study at one centre in Vienna, Austria. Healthy men and women aged 18-45 years with no comorbidities were randomly assigned, by computer-generated block randomisation (block size of 14), to receive either one of three escalating doses of the measles-virus-based candidate vaccine (low dose [1·5 × 10(4) median tissue culture infection doses (TCID50) per 0·05 mL], medium dose [7·5 × 10(4) TCID50 per 0·25 mL], or high dose [3·0 × 10(5) TCID50 per 1·0 mL]), or the active comparator-Priorix. Participants were additionally block-randomised to receive a booster injection on either day 28 or day 90 after the first vaccination. Participants and study investigators were masked to group allocation. The primary endpoint was the presence of neutralising anti-chikungunya antibodies on day 28, as assessed by 50% plaque reduction neutralisation test. Analysis was by intention to treat and per protocol. This trial is registered with EudraCT, number 2013-001084-23. FINDINGS: Between Nov 22, 2013, and Feb 25, 2014, we randomly assigned 42 participants to receive the low dose (n=12), the medium dose (n=12), or the high dose (n=12) of the measles-virus-based candidate vaccine, or Priorix (n=6), of whom 36 participants (86%; n=9, n=12, n=10, n=5, respectively) were included in the per-protocol population. The candidate vaccine raised neutralising antibodies in all dose cohorts after one immunisation, with seroconversion rates of 44% (n=4) in the low-dose group, 92% (n=11) in the medium-dose group, and 90% (n=10) in the high-dose group. The immunogenicity of the candidate vaccine was not affected by pre-existing anti-measles immunity. The second vaccination resulted in a 100% seroconversion for all participants in the candidate vaccine groups. The candidate vaccine had an overall good safety profile, and the rate of adverse events increased with vaccine dose and volume. No vaccination-related serious adverse events were recorded. INTERPRETATION: The live recombinant measles-virus-based chikungunya vaccine had good immunogenicity, even in the presence of anti-vector immunity, was safe, and had a generally acceptable tolerability profile. This vaccine is the first promising measles-virus-based candidate vaccine for use in human beings. FUNDING: Themis Bioscience GmBH.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/administração & dosagem , Febre de Chikungunya/prevenção & controle , Vírus Chikungunya/imunologia , Vírus do Sarampo/imunologia , Vacinas Virais/administração & dosagem , Adolescente , Adulto , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Antígenos Virais/imunologia , Febre de Chikungunya/imunologia , Febre de Chikungunya/virologia , Vírus Chikungunya/genética , Método Duplo-Cego , Esquema de Medicação , Feminino , Voluntários Saudáveis , Humanos , Masculino , Vírus do Sarampo/genética , Vacina contra Sarampo-Caxumba-Rubéola/administração & dosagem , Vacina contra Sarampo-Caxumba-Rubéola/imunologia , Pessoa de Meia-Idade , Vacinas Sintéticas , Vacinas Virais/imunologia
6.
Clin Vaccine Immunol ; 22(5): 516-25, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25761459

RESUMO

The envelope (E) protein of flaviviruses includes three domains, EI, EII, and EIII, and is the major protective antigen. Because EIII is rich in type-specific and subcomplex-specific neutralizing epitopes and is easy to express, it is particularly attractive as a recombinant vaccine antigen. VaxInnate has developed a vaccine platform that genetically links vaccine antigens to bacterial flagellin, a Toll-like receptor 5 ligand. Here we report that tetravalent dengue vaccines (TDVs) consisting of four constructs, each containing two copies of EIII fused to flagellin (R3.2x format), elicited robust and long-lived neutralizing antibodies (geometric mean titers of 200 to 3,000), as measured with a 50% focus reduction neutralization test (FRNT50). In an immunogenicity study, rhesus macaques (n = 2) immunized subcutaneously with 10 µg or 90 µg of TDV three or four times, at 4- to 6-week intervals, developed neutralizing antibodies to four dengue virus (DENV) serotypes (mean post-dose 3 FRNT50 titers of 102 to 601). In an efficacy study, rhesus macaques (n = 4) were immunized intramuscularly with 16 µg or 48 µg of TDV or a placebo control three times, at 1-month intervals. The animals that received 48-µg doses of TDV developed neutralizing antibodies against the four serotypes (geometric mean titers of 49 to 258) and exhibited reduced viremia after DENV-2 challenge, with a group mean viremia duration of 1.25 days and 2 of 4 animals being completely protected, compared to the placebo-treated animals, which all developed viremia, with a mean duration of 4 days. In conclusion, flagellin-EIII fusion vaccines are immunogenic and partially protective in a nonhuman primate model.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Citocinas/sangue , Citocinas/imunologia , Vacinas contra Dengue/administração & dosagem , Vírus da Dengue/genética , Modelos Animais de Doenças , Flagelina/administração & dosagem , Flagelina/imunologia , Injeções Subcutâneas , Macaca mulatta/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Receptor 5 Toll-Like/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Viremia/prevenção & controle
7.
Virus Genes ; 46(2): 203-18, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22411105

RESUMO

The prevalence of all four dengue virus (DENV) serotypes has increased dramatically in recent years in many tropical and sub-tropical countries accompanied by an increase in genetic diversity within each serotype. This expansion in genetic diversity is expected to give rise to viruses with altered antigenicity, virulence, and transmissibility. We previously demonstrated the co-circulation of multiple DENV genotypes in Thailand and identified a predominant genotype for each serotype. In this study, we performed a comparative analysis of the complete genomic sequences of 28 DENV-3 predominant genotype II strains previously collected during different DENV-3 epidemics in Thailand from 1973 to 2001 with the goal to define mutations that might correlate with virulence, transmission frequency, and epidemiological impact. The results revealed (1) 37 amino acid and six nucleotide substitutions adopted and fixed in the virus genome after their initial substitutions over nearly 30-year-sampling period, (2) the presence of more amino acid and nucleotide substitutions in recent virus isolates compared with earlier isolates, (3) six amino acid substitutions in capsid (C), pre-membrane (prM), envelope (E), and nonstructural (NS) proteins NS4B and NS5, which appeared to be associated with periods of high DENV-3 epidemic activity, (4) the highest degree of conservation in C, NS2B and the 5'-untranslated region (UTR), and (5) the highest percentage of amino acid substitutions in NS2A protein.


Assuntos
Vírus da Dengue/genética , Vírus da Dengue/isolamento & purificação , Dengue/virologia , Substituição de Aminoácidos , Dengue/epidemiologia , Vírus da Dengue/classificação , Genótipo , Humanos , Dados de Sequência Molecular , Filogenia , Tailândia/epidemiologia , Proteínas Virais/genética
8.
Am J Trop Med Hyg ; 88(1): 73-88, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23208878

RESUMO

Two formulations of a new live tetravalent dengue virus (DENV) vaccine produced using re-derived master seeds from a precursor vaccine and that same precursor vaccine as a control were compared in a placebo-controlled, randomized, observer-blind, phase II trial of 86 healthy adults. Two vaccine doses were administered 6 months apart; a third dose was offered to a subset. Symptoms and signs of dengue-like illness reported after vaccination were mild to moderate, transient, and occurred with similar frequency among recipients of the new DENV vaccine and placebo, except for rash. Neither dengue nor vaccine-related serious adverse events were reported. The first DENV vaccine dose was moderately immunogenic; the second dose increased the potency and breadth of the neutralizing antibody response. Tetravalent response rates to the new formulations were 60% and 66.7% in unprimed subjects. A third dose did not increase tetravalent antibody rates. The new DENV vaccine candidates merit additional evaluation.


Assuntos
Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/imunologia , Adulto , Vacinas contra Dengue/efeitos adversos , Vacinas contra Dengue/imunologia , Humanos , Placebos
9.
J Infect Dis ; 204(3): 442-50, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21742844

RESUMO

BACKGROUND: Live, multivalent vaccines have historically exhibited interference in humans; live dengue virus (DENV) vaccines have proven no exception. METHODS: To characterize interactions between DENV serotypes in a tetravalent live-attenuated virus vaccine candidate, we analyzed data from a factorial clinical trial in which all combinations of high- and low-dose DENV serotypes were combined in 16 live-attenuated tetravalent vaccine formulations (N = 64) and administered to flavivirus-naive adult volunteers. Regression models considered the outcomes of reactogenicity and seroconversion, controlling for all serotype doses simultaneously. Additionally, results were compared against earlier evaluations of the same viruses administered as monovalent formulations. RESULTS: DENV-1 was immunologically dominant in both monovalent and tetravalent formulations. In tetravalent formulations, DENV-1 and DENV-2 antagonized each other, with a high dose of one decreasing seroconversion to the other. However, high-dose DENV-1 significantly increased seroconversion against 3 or more serotypes, increasing seroconversion to DENV-1, DENV-3, and DENV-4. The highest reactogenicity occurred when DENV-1 was at high dose and all others were low; reactogenicity decreased with the incorporation of other high-dose serotypes. CONCLUSIONS: Interference and facilitation occurred between serotypes in the live vaccine candidate evaluated. These analyses suggest that it may be possible to exploit facilitation to increase overall seroconversion.


Assuntos
Vacinas contra Dengue/administração & dosagem , Vírus da Dengue/classificação , Anticorpos Antivirais/sangue , Ensaios Clínicos como Assunto , Vacinas contra Dengue/efeitos adversos , Vacinas contra Dengue/imunologia , Relação Dose-Resposta Imunológica , Humanos , Modelos Logísticos , Sorotipagem , Vacinação/efeitos adversos
10.
Vaccine ; 29(35): 5925-31, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21723353

RESUMO

Immune sera from volunteers vaccinated in a blinded Phase 3 clinical trial with JE-VAX(®) and a new Japanese encephalitis virus (JEV) vaccine (IC51 or IXIARO), were tested for the ability to protect mice against lethal JEV challenge. Sera from IXIARO vaccinated subjects were pooled into four batches based on neutralizing antibody measured by plaque reduction neutralization test (PRNT(50) titer): high (∼200), medium (∼40-50), low (∼20) and negative (<10). Pooled sera from JE-VAX(®) vaccinated subjects (PRNT(50) titer∼55) and pooled JEV antibody negative pre-vaccination sera were used as controls. Groups of ten 6- to 7-week-old female ICR mice were injected intraperitoneally with 0.5 ml of each serum pool diluted 1:2 or 1:10, challenged approximately 18 h later with a lethal dose of either JEV strain SA14 (genotype III) or strain KE-093 (genotype I) and observed for 21 days. All mice in the non-immune serum groups developed clinical signs consistent with JEV infection or died, whereas high titer sera from both IXIARO and JE-VAX(®) sera protected 90-100% of the animals. Statistical tests showed similar protection against both JEV strains SA14 and KE-093 and protection correlated with the anti-JEV antibody titer of IXIARO sera as measured by PRNT(50). Ex vivo neutralizing antibody titers showed that almost all mice with a titer of 10 or greater were fully protected. In a separate study, analysis of geometric mean titers (GMTs) of the groups of mice vaccinated with different doses of IXIARO and challenged with JEV SA14 provided additional evidence that titers≥10 were protective.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/prevenção & controle , Vacinas contra Encefalite Japonesa/administração & dosagem , Animais , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Encefalite Japonesa/imunologia , Encefalite Japonesa/mortalidade , Encefalite Japonesa/virologia , Feminino , Humanos , Soros Imunes/administração & dosagem , Soros Imunes/imunologia , Imunização , Imunização Passiva , Vacinas contra Encefalite Japonesa/imunologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Testes de Neutralização , Análise de Sobrevida , Resultado do Tratamento
11.
Clin Vaccine Immunol ; 18(4): 523-32, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21270280

RESUMO

Dengue is a mosquito-borne infection caused by four distinct serotypes of dengue virus, each appearing cyclically in the tropics and subtropics along the equator. Although vaccines are currently under development, none are available to the general population. One of the main impediments to the successful advancement of these vaccines is the lack of well-defined immune correlates of protection. Here, we describe a protein microarray approach for measuring antibody responses to the complete viral proteome comprised of the structural (capsid, membrane, and envelope) and nonstructural (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) components of all four dengue virus serotypes (1 to 4). We examined rhesus macaques vaccinated with tetravalent vaccines consisting of live-attenuated virus (LAV) or purified inactivated virus (PIV), followed by boosting with LAV and challenging with wild-type dengue virus. We detected temporal increases in antibodies against envelope proteins in response to either vaccine, while only the PIV/LAV vaccination strategy resulted in anticapsid antibodies. In contrast to results from vaccination, naïve macaques challenged with wild-type viruses of each serotype demonstrated a balanced response to nonstructural and structural components, including responses against the membrane protein. Our results demonstrate discriminating details concerning the nature of antibody responses to dengue virus at the proteomic level and suggest the usefulness of this information for vaccine development.


Assuntos
Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Proteínas Virais/imunologia , Animais , Macaca mulatta , Análise em Microsséries , Análise Serial de Proteínas/métodos
12.
Virus Genes ; 42(2): 200-3, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21234665

RESUMO

Eight serum specimens collected from dengue patients in Guatemala and Honduras during the Central American epidemic of 2007 were analyzed. Virus identification and serotyping performed by a nested RT-PCR assay revealed two DENV-1 isolates from Guatemala, four DENV-2 isolates, two each from Guatemala and Honduras, and two DENV-4 isolates from Honduras. Viral genotyping determined by phylogenetic analysis of the complete envelope gene sequences demonstrated that the DENV-2 isolates from Guatemala and Honduras fell into the American/Asian Genotype III, and were most closely related to DENV-2/NI/BID-V2683-1999 isolated from a dengue case in Nicaragua in 1999; and the DENV-4 F07-076 isolate from Honduras belonged to genotype II, and was most closely related to DENV-4/US/BID-V1093/1998 isolated from Puerto Rico in 1998. Our results suggest that the 2007 dengue outbreaks in Guatemala and Honduras were most likely caused by the re-emergence of earlier, indigenous DENV strains rather than by newly introduced strains and there were at least three serotypes of DENV co-circulating during the 2007 Central American epidemics.


Assuntos
Vírus da Dengue/genética , Dengue/virologia , Epidemias , Proteínas do Envelope Viral/genética , Dengue/epidemiologia , Dengue/genética , Genótipo , Guatemala/epidemiologia , Honduras/epidemiologia , Humanos , Filogenia , RNA Viral/genética
13.
Am J Trop Med Hyg ; 83(5): 1156-65, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21036855

RESUMO

Comparative sequence analysis was performed on the full-length genomic sequences of 10 representative dengue virus serotype 1 (DENV-1) strains sampled from patients at Children's Hospital, Bangkok, Thailand over a 22-year period, which represented different epidemics, disease severity, and sampling time. The results showed remarkable inter-genotypic variation between predominant and non-predominant genotypes and genotype-specific amino acids and nucleotides throughout the entire viral genome except for the 5'-non-translated region. The frequency of intra-genotypic variation was correlated with dengue transmission rate and sampling time. The 5'-non-translated region of all 10 viruses was highly conserved for predominant and non-predominant genotypes and NS2B was the most conserved protein. Some intra-genotypic substitutions of amino acids and nucleotides in predominant genotype strains were fixed in the viral genome since 1994, which indicated that the evolution of predominant genotype strains in situ over time might contribute to increased virus fitness important for sustaining dengue epidemics in Thailand.


Assuntos
Vírus da Dengue/classificação , Vírus da Dengue/genética , Dengue/epidemiologia , Dengue/virologia , Surtos de Doenças , Substituição de Aminoácidos , Sequência de Bases , Criança , Pré-Escolar , Vírus da Dengue/patogenicidade , Feminino , Genoma Viral , Genótipo , Humanos , Lactente , Masculino , RNA Viral/química , Índice de Gravidade de Doença , Tailândia/epidemiologia , Fatores de Tempo
14.
Virus Genes ; 41(1): 5-13, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20336481

RESUMO

In order to characterize viral genetic variation among predominant and non-predominant genotypes of Thai dengue serotype 4 viruses (DENV-4) and follow mutations that occur during virus evolution, we performed a comparative analysis of the complete genomic sequences of six DENV-4 isolates representing three genotypes (I, IIA, and III) co-circulating in Thailand over a 24-year period. The results revealed [1] remarkable genetic variation in the viral genome between predominant and non-predominant genotypes; [2] inter-genotype-specific amino acid and nucleotide mutations in most regions of the viral genome; [3] more amino acid and nucleotide substitutions in later as compared to earlier isolates for predominant genotype I strains; [4] a single nucleotide substitution at nucleotide position 77 of the 5-'NTR of two non-predominant genotype III strains that disrupted a small conserved 3'stem-loop (SL) in the cyclization sequence required for virus replication; [5] a high degree of conservation of PrM/M and NS2B proteins, and the 5'-NTR in predominant genotype I strains with no mutations observed over the 24-year period of observation; and [6] no molecular markers that appeared to correlate with disease severity. Several mutations identified in this study might have a significant impact on the persistence of virus in the population, including one in the 5'-NTR that disrupted a small, highly conserved 3'SL2 structure at the terminus of the cyclized 5'-3' RNA sequences in two genotype III strains, and three amino acid (aa) charge change mutations in the E and NS5 proteins of genotype I strains. The conserved 3'-SL structure may be a target for antiviral drug development.


Assuntos
Vírus da Dengue/genética , Dengue/virologia , Mutação , Adolescente , Sequência de Bases , Criança , Pré-Escolar , Vírus da Dengue/isolamento & purificação , Feminino , Genótipo , Humanos , Masculino , Conformação de Ácido Nucleico , Sorotipagem , Tailândia , Fatores de Tempo
15.
Virology ; 396(2): 280-8, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19913867

RESUMO

A new vaccination strategy for dengue virus (DENV) was evaluated in rhesus macaques by priming with tetravalent purified inactivated virus (TPIV) or tetravalent plasmid DNA vaccines expressing the structural prME gene region (TDNA) then boosting 2 months later with a tetravalent live attenuated virus (TLAV) vaccine. Both vaccine combinations elicited virus neutralizing (N) antibodies. The TPIV/TLAV combination afforded complete protection against DENV 3 challenge at month 8. In a second experiment, priming with TPIV elicited N antibodies against all four serotypes (GMT 1:28 to 1:43). Boosting with TLAV led to an increase in the GMT for each serotype (1:500 to 1:1200 for DENVs 1, 3, and 4, and greater than 1:6000 for DENV 2), which declined by month 8 (GMT 1:62 for DENV 3, 1:154 for DENV 1, 1:174 for DENV 4, and 1:767 for DENV 2). After challenge with each one of the four DENV serotypes, vaccinated animals exhibited no viremia but showed anamnestic antibody responses to the challenge viruses.


Assuntos
Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Animais , Formação de Anticorpos/imunologia , Dengue/imunologia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/imunologia , Modelos Animais de Doenças , Humanos , Imunização Secundária , Macaca mulatta/imunologia , Macaca mulatta/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico , Viremia/imunologia
16.
Am J Trop Med Hyg ; 81(5): 825-33, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19861618

RESUMO

Dengue virus (DENV) infection is a worsening global health problem. The plaque reduction neutralization test (PRNT) is currently considered to be the "gold standard" to characterize and quantify circulating levels of anti-DENV neutralizing antibody (NAb). Many variations of the PRNT are currently in use and neither the assay nor its performance conditions have been standardized or harmonized between laboratories. We used a well-characterized panel of acute and late convalescent follow-up sera samples from children experiencing primary and secondary DENV infections to evaluate the performance of the dengue PRNT under a variety of testing conditions. Investigators varied cell type, control virus passage, and the use of complement across multiple assay runs of the same sample panel. Our findings indicate wide variation in PRNT titer results in response to varied testing conditions.


Assuntos
Dengue/diagnóstico , Dengue/epidemiologia , Testes de Neutralização/métodos , Ensaio de Placa Viral/métodos , Adolescente , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , Criança , Pré-Escolar , Cricetinae , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos
17.
Virus Res ; 145(1): 31-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19540887

RESUMO

Eighteen dengue viruses (DENVs) representing all four serotypes, isolated from pediatric patients at children's hospital, Queen Sirikit National Institute of Child Health, Bangkok, Thailand exhibiting a diverse spectrum of disease ranging from uncomplicated dengue fever (DF) to severe dengue hemorrhagic fever (DHF), were tested for their ability to attach to host cells, replicate and interfere with the IFNalpha signaling pathway by interfering with signal transducer and activator of transcription 1 (STAT-1) function. Although most isolates suppressed IFNalpha-induced STAT-1 phosphorylation, our results showed no difference between DENV strains associated with DF and those associated with DHF. However, the DHF isolates tended replicate to higher titers in dendritic cells (DCs) than the DF isolates, but this ability was independent of their cell-binding capability. Our results suggest that the emergence early in infection of viruses with a high degree of replication fitness may play an important role in DENV pathogenesis.


Assuntos
Vírus da Dengue , Interferon-alfa/metabolismo , Dengue Grave/virologia , Ligação Viral , Replicação Viral , Adolescente , Linhagem Celular , Criança , Pré-Escolar , Vírus da Dengue/isolamento & purificação , Vírus da Dengue/fisiologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Lactente , Masculino , Fator de Transcrição STAT1/metabolismo , Dengue Grave/metabolismo , Transdução de Sinais , Tailândia
18.
Vaccine ; 25(17): 3445-53, 2007 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-17241714

RESUMO

Japanese encephalitis (JE) is a serious disease caused by the JE virus. New generation JE vaccines are needed to prevent this disease. We conducted this Phase 2 randomized, open label, unblinded, single center study of a new, cell-culture derived, purified inactivated virus (JE-PIV) vaccine. The JE-PIV vaccine was administered in either two or three intramuscular (IM) doses (6.0 or 12.0 mcg each) with observation over 8 weeks. All volunteers completed the protocol without serious adverse reactions. Headache and transient tenderness at the injection site were the most common complaints. There were no laboratory abnormalities believed to be related to vaccine during the study. JE-PIV was well tolerated, resulted in high seroconversion rates [Day 56 (primary endpoint); 95-100%] and induced enduring immune responses up to 2 years after vaccination. Expanded Phase 3 trials are planned.


Assuntos
Vacinas contra Encefalite Japonesa/imunologia , Adulto , Anticorpos Antivirais/sangue , Encefalite Japonesa/prevenção & controle , Feminino , Humanos , Vacinas contra Encefalite Japonesa/efeitos adversos , Masculino , Vacinação , Vacinas de Produtos Inativados/imunologia
19.
J Virol ; 80(19): 9577-85, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973561

RESUMO

We evaluated three nonreplicating dengue virus type 2 (DENV-2) vaccines: (i) a DNA vaccine containing the prM-E gene region (D), (ii) a recombinant subunit protein vaccine containing the B domain (i.e., domain III) of the E protein as a fusion with the Escherichia coli maltose-binding protein (R), and (iii) a purified inactivated virus vaccine (P). Groups of four rhesus macaques each were primed once and boosted twice using seven different vaccination regimens. After primary vaccination, enzyme-linked immunosorbent assay (ELISA) antibody levels increased most rapidly for groups inoculated with the P and DP combination, and by 1 month after the second boost, ELISA titers were similar for all groups. The highest plaque reduction neutralization test (PRNT) titers were seen in those groups that received the DR/DR/DR combination (geometric mean titer [GMT], 510), the P/P/P vaccine (GMT, 345), the DP/DP/DP combination (GMT, 287), and the R/R/R vaccine (GMT, 200). The next highest titers were seen in animals that received the D/R/R vaccine (GMT, 186) and the D/P/P vaccine (GMT, 163). Animals that received the D/D/D vaccine had the lowest neutralizing antibody titer (GMT, 49). Both ELISA and PRNT titers declined at variable rates. The only significant protection from viremia was observed in the P-vaccinated animals (mean of 0.5 days), which also showed the highest antibody concentration, including antibodies to NS1, and highest antibody avidity at the time of challenge.


Assuntos
Anticorpos Antivirais/imunologia , Vírus da Dengue/imunologia , Vacinas de DNA/imunologia , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/farmacologia , Afinidade de Anticorpos/imunologia , Chlorocebus aethiops , Vírus da Dengue/classificação , Memória Imunológica/imunologia , Macaca mulatta , Células Vero , Viremia/imunologia , Viremia/prevenção & controle , Viremia/virologia
20.
Am J Trop Med Hyg ; 68(4): 469-76, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12875299

RESUMO

A nucleic acid vaccine for dengue-2 virus was developed, consisting of a plasmid DNA vector with the pre-membrane (prM) and envelope (E) genes expressed from a cytomegalovirus promoter. The DNA was adsorbed onto gold microspheres for administration by a gene gun. Expression was demonstrated by transfection of mouse cells in culture where the prM and E antigens were detected intracellularly, and the E antigen was detected in the culture supernatant fluid, similar to a natural infection. The vaccine elicited neutralizing antibodies to dengue-2 virus and antigen-specific cytotoxic T lymphocyte responses in mice. Several vaccination regimens were evaluated in rhesus macaques for the ability to elicit neutralizing antibodies and protect against viremia after challenge with live dengue-2 virus. Neutralizing antibodies were measured in three of three animals that received four 2-microg doses of DNA and in two of six animals that received two 1-microg doses. No antibodies were detected in three animals that received a single 1-microg dose. When dengue virus challenge was performed one month after vaccination, the three animals that received four 2-microg doses exhibited 0, 0, and 1 day of viremia compared with unimmunized controls which exhibited 4, 4, and 6 days of viremia. Three animals that received two 1-microg doses also exhibited 0, 0, and 1 day of viremia, whereas three animals that received a single 1-microg dose exhibited 2, 3, and 5 days of viremia compared with unimmunized controls, which exhibited 4 days of viremia each. When challenge was performed 7 months after vaccination, three animals that received two 1-microg doses exhibited 0, 3, and 5 days of viremia compared with unimmunized controls, which exhibited 4, 5, and 9 days of viremia. These results suggest that a regimen consisting of two 1-microg doses of DNA can confer satisfactory protection at one month, but not at seven months, after vaccination. Long-term protection following DNA vaccination may require revaccination, higher doses of DNA, or a vaccine that contains additional epitopes or adjuvants.


Assuntos
Vírus da Dengue/imunologia , Dengue/prevenção & controle , Vacinação/métodos , Vacinas de DNA , Vacinas Virais , Animais , Anticorpos Antivirais/sangue , Biolística , Vírus da Dengue/genética , Relação Dose-Resposta Imunológica , Feminino , Regulação Viral da Expressão Gênica , Vetores Genéticos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microesferas , Testes de Neutralização , Plasmídeos , Linfócitos T Citotóxicos/imunologia , Fatores de Tempo , Vacinas de DNA/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas da Matriz Viral/genética , Vacinas Virais/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA